1
|
Sharma A, Rudrawar S, Bharate SB, Jadhav HR. Recent advancements in the therapeutic approaches for Alzheimer's disease treatment: current and future perspective. RSC Med Chem 2025; 16:652-693. [PMID: 39790124 PMCID: PMC11707861 DOI: 10.1039/d4md00630e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 12/05/2024] [Indexed: 01/12/2025] Open
Abstract
Alzheimer's disease (AD) is a complex, incurable neurological condition characterized by cognitive decline, cholinergic neuron reduction, and neuronal loss. Its exact pathology remains uncertain, but multiple treatment hypotheses have emerged. The current treatments, single or combined, alleviate only symptoms and struggle to manage AD due to its multifaceted pathology. The developmental drugs target pivotal disease factors involved in the envisaged hypotheses and include targets such as amyloid aggregation, hyperphosphorylated tau proteins, and receptors like cholinergic, adrenergic, etc. Present-day research focuses on multi-target directed ligands (MTDLs), which inhibit multiple factors simultaneously, helping slow the disease's progression. This review attempts to collate the recent information related to proposed hypotheses for AD etiology. It systematically organizes the advances in various therapeutic options for AD, with a particular emphasis on clinical candidates. Also, it is expected to help medicinal chemists design novel AD treatments based on available information, which could be helpful to AD patients.
Collapse
Affiliation(s)
- Amit Sharma
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Sciences Pilani Pilani Campus, Vidya Vihar Pilani 333031 RJ India +91 1596 244183 +91 1596 255 506
| | - Santosh Rudrawar
- The Institute for Biomedicine and Glycomics, Griffith University Gold Coast 4222 Australia
- School of Pharmacy and Medical Sciences, Griffith University Gold Coast 4222 Australia
| | - Sandip B Bharate
- Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine Canal Road Jammu 181110 India
| | - Hemant R Jadhav
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Sciences Pilani Pilani Campus, Vidya Vihar Pilani 333031 RJ India +91 1596 244183 +91 1596 255 506
| |
Collapse
|
2
|
Ait Hamdan Y, El-Mansoury B, Elouali S, Rachmoune K, Belbachir A, Oudadesse H, Rhazi M. A review of chitosan polysaccharides: Neuropharmacological implications and tissue regeneration. Int J Biol Macromol 2024; 279:135356. [PMID: 39244136 DOI: 10.1016/j.ijbiomac.2024.135356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 08/20/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024]
Abstract
One of the current challenges in targeting neurological disorders is that many therapeutic molecules cannot cross the blood-brain barrier (BBB), which limits the use of natural molecules in nervous tissue regeneration. Thus, the development of new drugs to effectively treat neurological disorders would be a challenge. Natural resources are well known as a source of several therapeutic agents for the treatment of neurologic disorders. Recently, chitosan (CTS) and its derivatives from arthropod exoskeletons, have attracted much attention as a drug delivery system to transport therapeutic substances across the BBB and thanks to other neuroprotective effects including the participation to the CNS regenerations scaffolds to replicate the extracellular matrix and microenvironment of the body. This review will discuss the place of natural resource therapy in targeting neurological disorders. In particular, it will highlight recent understanding and progress in the applications of CTS as drug delivery systems and their therapeutic effects on these disorders through tissue regeneration, as well as the molecular mechanisms by which they exert these effects.
Collapse
Affiliation(s)
- Youssef Ait Hamdan
- Interdisciplinary Laboratory of Research in Bio-Resources, Environment and Materials, Higher Normal School, Cadi Ayyad University, 40000 Marrakech, Morocco; Univ Rennes, CNRS, ISCR-UMR 6226, F-35000 Rennes, France.
| | - Bilal El-Mansoury
- Laboratory of Anthropogenic, Biotechnology and Health, Team physiopathology Nutritional, Neurosciences and Toxicology, Faculty of Sciences, Chouaib Doukkali University, Av. Des facultés, 24000 El Jadida, Morocco
| | - Samia Elouali
- Interdisciplinary Laboratory of Research in Bio-Resources, Environment and Materials, Higher Normal School, Cadi Ayyad University, 40000 Marrakech, Morocco; University of Mons (UMONS) - Laboratory of Polymeric and Composite Materials (LPCM), Center of Innovation and Research in Materials and Polymers (CIRMAP), Place du Parc 20, 7000 Mons, Belgium
| | - Khawla Rachmoune
- Interdisciplinary Laboratory of Research in Bio-Resources, Environment and Materials, Higher Normal School, Cadi Ayyad University, 40000 Marrakech, Morocco; Biotechnology and Biomolecule Engineering Unit, CNESTEN, Rabat, Morocco
| | - Anass Belbachir
- Center for Regenerative Medicine, CHU MOHAMMED VI, Marrakech, Morocco; Faculty of Medicine and Pharmacy, Cadi Ayyad University, Marrakech, Morocco
| | | | - Mohammed Rhazi
- Interdisciplinary Laboratory of Research in Bio-Resources, Environment and Materials, Higher Normal School, Cadi Ayyad University, 40000 Marrakech, Morocco
| |
Collapse
|
3
|
Ghosh AK. BACE1 inhibitor drugs for the treatment of Alzheimer's disease: Lessons learned, challenges to overcome, and future prospects †. Glob Health Med 2024; 6:164-168. [PMID: 38947412 PMCID: PMC11197157 DOI: 10.35772/ghm.2024.01033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 06/04/2024] [Indexed: 07/02/2024]
Abstract
Alzheimer's disease (AD), first diagnosed over a century ago, remains one of the major healthcare crises around the globe. Currently, there is no cure or effective treatment. The majority of drug development efforts to date have targeted reduction of amyloid-β peptide (Aβ). Drug development through inhibition of beta-site amyloid precursor protein cleaving enzyme 1 (BACE1), resulted in promising early clinical studies. However, nearly all small molecule BACE1 inhibitor drugs failed to live up to expectations in later phase clinical trials, due to toxicity and efficacy issues. This commentary aims to provide a brief review of over two decades of BACE1 inhibitor drug development challenges and efforts for treatment of AD and prospects of future BACE1-based drugs.
Collapse
Affiliation(s)
- Arun K. Ghosh
- Departments of Chemistry, Purdue University, West Lafayette, IN, USA
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
4
|
Bernard PJ, Bellili D, Ismaili L. Calcium channel blockers' contribution to overcoming Current drug discovery challenges in Alzheimer's disease. Expert Opin Drug Discov 2024; 19:21-32. [PMID: 37800853 DOI: 10.1080/17460441.2023.2266994] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 10/02/2023] [Indexed: 10/07/2023]
Abstract
INTRODUCTION Alzheimer's disease (AD) is a progressive, irreversible, and multifactorial brain disorder that gradually and insidiously destroys individual's memory, thinking, and other cognitive abilities. AREAS COVERED In this perspective, the authors examine the complex and multifactorial nature of Alzheimer's disease and believe that the best approach to develop new drugs is the MTDL strategy, which obviously faces several challenges. These challenges include identifying the key combination of targets and their suitability for coordinated actions, as well as developing an acceptable pharmacokinetic and toxicological profile to deliver a drug candidate. EXPERT OPINION Since calcium plays a crucial role in the pathology of AD, a polypharmacological approach with calcium channel blockers reinforced by activities targeting other factors involved in AD is a serious option in our opinion. This is exemplified by a phase III clinical trial using a drug combination approach with Losartan, Amlodipine (a calcium channel blocker), and Atorvastatin, as well as several MTDL-based calcium channel blockade approaches with a promising in vitro and in vivo profile.
Collapse
Affiliation(s)
- Paul J Bernard
- Université de Franche-Comté, LINC, UFR Santé, Pôle de Chimie Médicinale, Besançon, France
| | - Djamila Bellili
- Université de Franche-Comté, LINC, UFR Santé, Pôle de Chimie Médicinale, Besançon, France
| | - Lhassane Ismaili
- Université de Franche-Comté, LINC, UFR Santé, Pôle de Chimie Médicinale, Besançon, France
| |
Collapse
|
5
|
Yeap YJ, Kandiah N, Nizetic D, Lim KL. BACE2: A Promising Neuroprotective Candidate for Alzheimer's Disease. J Alzheimers Dis 2023; 94:S159-S171. [PMID: 36463454 PMCID: PMC10473127 DOI: 10.3233/jad-220867] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2022] [Indexed: 11/30/2022]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia that affects millions of predominantly elderly individuals worldwide. Despite intensive research over several decades, controversies still surround the etiology of AD and the disease remains incurable. Meanwhile, new molecular players of the central amyloid cascade hypothesis have emerged and among these is a protease known as β-site APP cleavage enzyme 2 (BACE2). Unlike BACE1, BACE2 cleaves the amyloid-β protein precursor within the Aβ domain that accordingly prevents the generation of Aβ42 peptides, the aggregation of which is commonly regarded as the toxic entity that drives neurodegeneration in AD. Given this non-amyloidogenic role of BACE2, it is attractive to position BACE2 as a therapeutic target for AD. Indeed, several groups including ours have demonstrated a neuroprotective role for BACE2 in AD. In this review, we discuss emerging evidence supporting the ability of BACE2 in mitigating AD-associated pathology in various experimental systems including human pluripotent stem cell-derived cerebral organoid disease models. Alongside this, we also provide an update on the identification of single nucleotide polymorphisms occurring in the BACE2 gene that are linked to increased risk and earlier disease onset in the general population. In particular, we highlight a recently identified point mutation on BACE2 that apparently leads to sporadic early-onset AD. We believe that a better understanding of the role of BACE2 in AD would provide new insights for the development of viable therapeutic strategies for individuals with dementia.
Collapse
Affiliation(s)
- Yee Jie Yeap
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Nagaendran Kandiah
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Dean Nizetic
- Barts & The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Kah-Leong Lim
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Department of Brain Sciences, Imperial College London, London, UK
- Shanxi Medical University, Taiyuan, People’s Republic of China
| |
Collapse
|
6
|
Multicomponent reactions as a privileged tool for multitarget-directed ligand strategies in Alzheimer's disease therapy. Future Med Chem 2022; 14:1583-1606. [PMID: 36263996 DOI: 10.4155/fmc-2022-0170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Among neurodegenerative pathologies affecting the older population, Alzheimer's disease is the most common type of dementia and leads to neurocognitive and behavioral disorders. It is a complex and progressive age-related multifactorial disease characterized by a series of highly interconnected pathophysiological processes. Within the last decade, the multitarget-directed ligand strategy has emerged as a viable approach to developing complex molecules that exhibit several pharmacophores which can target the different enzymes and receptors involved in the pathogenesis of the disease. Herein, we focus on using multicomponent reactions such as Hantzsch, Biginelli and Ugi to develop these biologically active multitopic ligands.
Collapse
|
7
|
Alić I, Goh PA, Murray A, Portelius E, Gkanatsiou E, Gough G, Mok KY, Koschut D, Brunmeir R, Yeap YJ, O'Brien NL, Groet J, Shao X, Havlicek S, Dunn NR, Kvartsberg H, Brinkmalm G, Hithersay R, Startin C, Hamburg S, Phillips M, Pervushin K, Turmaine M, Wallon D, Rovelet-Lecrux A, Soininen H, Volpi E, Martin JE, Foo JN, Becker DL, Rostagno A, Ghiso J, Krsnik Ž, Šimić G, Kostović I, Mitrečić D, Francis PT, Blennow K, Strydom A, Hardy J, Zetterberg H, Nižetić D. Patient-specific Alzheimer-like pathology in trisomy 21 cerebral organoids reveals BACE2 as a gene dose-sensitive AD suppressor in human brain. Mol Psychiatry 2021; 26:5766-5788. [PMID: 32647257 PMCID: PMC8190957 DOI: 10.1038/s41380-020-0806-5] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 05/18/2020] [Accepted: 05/29/2020] [Indexed: 11/15/2022]
Abstract
A population of more than six million people worldwide at high risk of Alzheimer's disease (AD) are those with Down Syndrome (DS, caused by trisomy 21 (T21)), 70% of whom develop dementia during lifetime, caused by an extra copy of β-amyloid-(Aβ)-precursor-protein gene. We report AD-like pathology in cerebral organoids grown in vitro from non-invasively sampled strands of hair from 71% of DS donors. The pathology consisted of extracellular diffuse and fibrillar Aβ deposits, hyperphosphorylated/pathologically conformed Tau, and premature neuronal loss. Presence/absence of AD-like pathology was donor-specific (reproducible between individual organoids/iPSC lines/experiments). Pathology could be triggered in pathology-negative T21 organoids by CRISPR/Cas9-mediated elimination of the third copy of chromosome 21 gene BACE2, but prevented by combined chemical β and γ-secretase inhibition. We found that T21 organoids secrete increased proportions of Aβ-preventing (Aβ1-19) and Aβ-degradation products (Aβ1-20 and Aβ1-34). We show these profiles mirror in cerebrospinal fluid of people with DS. We demonstrate that this protective mechanism is mediated by BACE2-trisomy and cross-inhibited by clinically trialled BACE1 inhibitors. Combined, our data prove the physiological role of BACE2 as a dose-sensitive AD-suppressor gene, potentially explaining the dementia delay in ~30% of people with DS. We also show that DS cerebral organoids could be explored as pre-morbid AD-risk population detector and a system for hypothesis-free drug screens as well as identification of natural suppressor genes for neurodegenerative diseases.
Collapse
Grants
- MR/S011277/1 Medical Research Council
- MR/L501542/1 Medical Research Council
- RF1 AG059695 NIA NIH HHS
- G-0907 Parkinson's UK
- MR/N026004/1 Medical Research Council
- MR/R024901/1 Medical Research Council
- Wellcome Trust
- 217199 Wellcome Trust
- G0901254 Medical Research Council
- G0701075 Medical Research Council
- 098330 Wellcome Trust
- William Harvey Academy Fellowship, co-funded by the People Programme (Marie Curie Actions) of the European Union’s Seventh Framework Programme (FP7/2007-2013) under REA grant agreement n° 608765
- Fondation pour la Recherche Médicale (Foundation for Medical Research in France)
- National Research Foundation Singapore (National Research Foundation-Prime Minister’s office, Republic of Singapore)
- BrightFocus Foundation (BrightFocus)
- Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- Svenska Forskningsrådet Formas (Swedish Research Council Formas)
- KB holds the Torsten Söderberg Professorship in Medicine at the Royal Swedish Academy of Sciences, and is supported by the Swedish Alzheimer Foundation (#AF-742881), Hjärnfonden, Sweden (#FO2017-0243), and the Swedish State Support for Clinical Research (#ALFGBG-715986).
- Wellcome Trust (Wellcome)
- JH received funding from the Dementia Research Institute, an anonymous foundation and the Dolby foundation
- HZ is a Wallenberg Academy Fellow supported by grants from the Swedish Research Council, the European Research Council, Swedish State Support for Clinical Research (ALFGBG-720931) the UK Dementia Research Institute at UCL
Collapse
Affiliation(s)
- Ivan Alić
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore
- The Blizard Institute, Barts & The London School of Medicine, Queen Mary University of London, London, E1 2AT, UK
- Department of Anatomy, Histology and Embryology, Faculty of Veterinary Medicine, University of Zagreb, 10000, Zagreb, Croatia
| | - Pollyanna A Goh
- The Blizard Institute, Barts & The London School of Medicine, Queen Mary University of London, London, E1 2AT, UK
- LonDownS Consortium, London, UK
| | - Aoife Murray
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore
| | - Erik Portelius
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, S-405 30, Sweden
| | - Eleni Gkanatsiou
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, S-405 30, Sweden
| | - Gillian Gough
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore
| | - Kin Y Mok
- LonDownS Consortium, London, UK
- Dementia Research Institute & Reta Lila Weston Institute, Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - David Koschut
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore
| | - Reinhard Brunmeir
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore
| | - Yee Jie Yeap
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore
| | - Niamh L O'Brien
- The Blizard Institute, Barts & The London School of Medicine, Queen Mary University of London, London, E1 2AT, UK
- LonDownS Consortium, London, UK
| | - Jürgen Groet
- The Blizard Institute, Barts & The London School of Medicine, Queen Mary University of London, London, E1 2AT, UK
- LonDownS Consortium, London, UK
| | - Xiaowei Shao
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore
| | - Steven Havlicek
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore, 138672, Singapore
| | - N Ray Dunn
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore
- Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), Singapore, 138648, Singapore
| | - Hlin Kvartsberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, S-405 30, Sweden
| | - Gunnar Brinkmalm
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, S-405 30, Sweden
| | - Rosalyn Hithersay
- LonDownS Consortium, London, UK
- Division of Psychiatry, University College London, London, WC1E 6BT, UK
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, SE5 8AF, UK
| | - Carla Startin
- LonDownS Consortium, London, UK
- Division of Psychiatry, University College London, London, WC1E 6BT, UK
| | - Sarah Hamburg
- LonDownS Consortium, London, UK
- Division of Psychiatry, University College London, London, WC1E 6BT, UK
| | - Margaret Phillips
- School of Biological Sciences, Nanyang Technological University, Singapore, 639798, Singapore
| | - Konstantin Pervushin
- School of Biological Sciences, Nanyang Technological University, Singapore, 639798, Singapore
| | - Mark Turmaine
- Division of Biosciences, University College London, Gower Street, London, WC1E 6BT, UK
| | - David Wallon
- Normandie Univ, UNIROUEN, Inserm U1245 and Rouen University Hospital, Department of Neurology and CNR-MAJ, F 76000, Normandy Center for Genomic and Personalized Medicine, Rouen, France
| | - Anne Rovelet-Lecrux
- Normandie Univ, UNIROUEN, Inserm U1245 and Rouen University Hospital, Department of Neurology and CNR-MAJ, F 76000, Normandy Center for Genomic and Personalized Medicine, Rouen, France
| | - Hilkka Soininen
- University of Eastern Finland, Institute of Clinical Medicine/Neurology, Kuopio, FI-70211, Finland
| | - Emanuela Volpi
- School of Life Sciences, University of Westminster, London, W1W 6UW, UK
| | - Joanne E Martin
- The Blizard Institute, Barts & The London School of Medicine, Queen Mary University of London, London, E1 2AT, UK
| | - Jia Nee Foo
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore, 138672, Singapore
| | - David L Becker
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore
| | - Agueda Rostagno
- Department of Pathology & Department of Psychiatry, New York University School of Medicine, New York, NY, 10016, USA
| | - Jorge Ghiso
- Department of Pathology & Department of Psychiatry, New York University School of Medicine, New York, NY, 10016, USA
| | - Željka Krsnik
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, 10000, Zagreb, Croatia
| | - Goran Šimić
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, 10000, Zagreb, Croatia
| | - Ivica Kostović
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, 10000, Zagreb, Croatia
| | - Dinko Mitrečić
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, 10000, Zagreb, Croatia
| | - Paul T Francis
- Wolfson Centre for Age-Related Diseases, King's College London, London, SE1 1UL, UK
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, S-405 30, Sweden
| | - Andre Strydom
- LonDownS Consortium, London, UK
- Division of Psychiatry, University College London, London, WC1E 6BT, UK
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, SE5 8AF, UK
| | - John Hardy
- LonDownS Consortium, London, UK
- Dementia Research Institute & Reta Lila Weston Institute, Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, S-405 30, Sweden
- Dementia Research Institute & Reta Lila Weston Institute, Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Dean Nižetić
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore.
- The Blizard Institute, Barts & The London School of Medicine, Queen Mary University of London, London, E1 2AT, UK.
- LonDownS Consortium, London, UK.
| |
Collapse
|
8
|
Nozal V, García‐Rubia A, Cuevas EP, Pérez C, Tosat‐Bitrián C, Bartolomé F, Carro E, Ramírez D, Palomo V, Martínez A. From Kinase Inhibitors to Multitarget Ligands as Powerful Drug Leads for Alzheimer's Disease using Protein-Templated Synthesis. Angew Chem Int Ed Engl 2021; 60:19344-19354. [PMID: 34169618 PMCID: PMC8457121 DOI: 10.1002/anie.202106295] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/24/2021] [Indexed: 11/24/2022]
Abstract
Multitarget directed ligands (MTDLs) are arising as promising tools to tackle complex diseases. The main goal of this work is to create powerful modulating agents for neurodegenerative disorders. To achieve this aim, we have combined fragments that inhibit key protein kinases involved in the main pathomolecular pathways of Alzheimer's disease (AD) such as tau aggregation, neuroinflammation and decreased neurogenesis, whilst looking for a third action in beta-secretase (BACE1), responsible of β-amyloid production. We obtained well-balanced MTDLs with in vitro activity in three different relevant targets and efficacy in two cellular models of AD. Furthermore, computational studies confirmed how these compounds accommodate adequately into the long and rather narrow BACE1 catalytic site. Finally, we employed in situ click chemistry using BACE1 as protein template as a versatile synthetic tool that allowed us to obtain further MTDLs.
Collapse
Affiliation(s)
- Vanesa Nozal
- Structural and Chemical Biology DepartmentCentro de Investigaciones Biológicas-CSICRamiro de Maeztu 928040MadridSpain
| | - Alfonso García‐Rubia
- Structural and Chemical Biology DepartmentCentro de Investigaciones Biológicas-CSICRamiro de Maeztu 928040MadridSpain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED)Instituto de Salud Carlos III28031MadridSpain
| | - Eva P. Cuevas
- Structural and Chemical Biology DepartmentCentro de Investigaciones Biológicas-CSICRamiro de Maeztu 928040MadridSpain
| | - Concepción Pérez
- Instituto de Química Médica-CSIC)Juan de la Cierva 328006MadridSpain
| | - Carlota Tosat‐Bitrián
- Structural and Chemical Biology DepartmentCentro de Investigaciones Biológicas-CSICRamiro de Maeztu 928040MadridSpain
| | - Fernando Bartolomé
- Hospital Universitario 12 de Octubre Research Institute (imas12)Group of Neurodegenerative DiseasesJuan de la Cierva 328006MadridSpain
| | - Eva Carro
- Hospital Universitario 12 de Octubre Research Institute (imas12)Group of Neurodegenerative DiseasesJuan de la Cierva 328006MadridSpain
| | - David Ramírez
- Instituto de Ciencias BiomédicasUniversidad Autónoma de ChileLlano Subercaseaux2801—piso 6SantiagoChile
| | - Valle Palomo
- Structural and Chemical Biology DepartmentCentro de Investigaciones Biológicas-CSICRamiro de Maeztu 928040MadridSpain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED)Instituto de Salud Carlos III28031MadridSpain
| | - Ana Martínez
- Structural and Chemical Biology DepartmentCentro de Investigaciones Biológicas-CSICRamiro de Maeztu 928040MadridSpain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED)Instituto de Salud Carlos III28031MadridSpain
| |
Collapse
|
9
|
Nozal V, García‐Rubia A, Cuevas EP, Pérez C, Tosat‐Bitrián C, Bartolomé F, Carro E, Ramírez D, Palomo V, Martínez A. From Kinase Inhibitors to Multitarget Ligands as Powerful Drug Leads for Alzheimer's Disease using Protein‐Templated Synthesis. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202106295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Vanesa Nozal
- Structural and Chemical Biology Department Centro de Investigaciones Biológicas-CSIC Ramiro de Maeztu 9 28040 Madrid Spain
| | - Alfonso García‐Rubia
- Structural and Chemical Biology Department Centro de Investigaciones Biológicas-CSIC Ramiro de Maeztu 9 28040 Madrid Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED) Instituto de Salud Carlos III 28031 Madrid Spain
| | - Eva P. Cuevas
- Structural and Chemical Biology Department Centro de Investigaciones Biológicas-CSIC Ramiro de Maeztu 9 28040 Madrid Spain
| | - Concepción Pérez
- Instituto de Química Médica-CSIC) Juan de la Cierva 3 28006 Madrid Spain
| | - Carlota Tosat‐Bitrián
- Structural and Chemical Biology Department Centro de Investigaciones Biológicas-CSIC Ramiro de Maeztu 9 28040 Madrid Spain
| | - Fernando Bartolomé
- Hospital Universitario 12 de Octubre Research Institute (imas12) Group of Neurodegenerative Diseases Juan de la Cierva 3 28006 Madrid Spain
| | - Eva Carro
- Hospital Universitario 12 de Octubre Research Institute (imas12) Group of Neurodegenerative Diseases Juan de la Cierva 3 28006 Madrid Spain
| | - David Ramírez
- Instituto de Ciencias Biomédicas Universidad Autónoma de Chile Llano Subercaseaux 2801—piso 6 Santiago Chile
| | - Valle Palomo
- Structural and Chemical Biology Department Centro de Investigaciones Biológicas-CSIC Ramiro de Maeztu 9 28040 Madrid Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED) Instituto de Salud Carlos III 28031 Madrid Spain
| | - Ana Martínez
- Structural and Chemical Biology Department Centro de Investigaciones Biológicas-CSIC Ramiro de Maeztu 9 28040 Madrid Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED) Instituto de Salud Carlos III 28031 Madrid Spain
| |
Collapse
|
10
|
Liu Y, Cong L, Han C, Li B, Dai R. Recent Progress in the Drug Development for the Treatment of Alzheimer's Disease Especially on Inhibition of Amyloid-peptide Aggregation. Mini Rev Med Chem 2021; 21:969-990. [PMID: 33245270 DOI: 10.2174/1389557520666201127104539] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 08/25/2020] [Accepted: 09/14/2020] [Indexed: 11/22/2022]
Abstract
As the world 's population is aging, Alzheimer's disease (AD) has become a big concern since AD has started affecting younger people and the population of AD patients is increasing worldwide. It has been revealed that the neuropathological hallmarks of AD are typically characterized by the presence of neurotoxic extracellular amyloid plaques in the brain, which are surrounded by tangles of neuronal fibers. However, the causes of AD have not been completely understood yet. Currently, there is no drug to effectively prevent AD or to completely reserve the symptoms in the patients. This article reviews the pathological features associated with AD, the recent progress in research on the drug development to treat AD, especially on the discovery of natural product derivatives to inhibit Aβ peptide aggregation as well as the design and synthesis of Aβ peptide aggregation inhibitors to treat AD.
Collapse
Affiliation(s)
- Yuanyuan Liu
- Advanced Research Institute of Multidisciplinary Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Lin Cong
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Life Science, Beijing Institute of Technology, Beijing, 10081, China
| | - Chu Han
- Advanced Research Institute of Multidisciplinary Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Bo Li
- Advanced Research Institute of Multidisciplinary Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Rongji Dai
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Life Science, Beijing Institute of Technology, Beijing, 10081, China
| |
Collapse
|
11
|
Affiliation(s)
- Ann T Nguyen
- Department of Psychology, School of Behavioral Health, Loma Linda University, Loma Linda, CA, USA
| | - Grace J Lee
- Department of Psychology, School of Behavioral Health, Loma Linda University, Loma Linda, CA, USA
| |
Collapse
|
12
|
Costa GP, Baldinotti RSM, Fronza MG, Nascimento JER, Dias ÍFC, Sonego MS, Seixas FK, Collares T, Perin G, Jacob RG, Savegnago L, Alves D. Synthesis, Molecular Docking, and Preliminary Evaluation of 2-(1,2,3-Triazoyl)benzaldehydes As Multifunctional Agents for the Treatment of Alzheimer's Disease. ChemMedChem 2020; 15:610-622. [PMID: 32012463 DOI: 10.1002/cmdc.201900622] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 01/20/2020] [Indexed: 12/11/2022]
Abstract
We described here our results on the use of thiourea as a ligand in the copper catalysed azide-alkyne cycloaddition (CuAAC) of 2-azidobenzaldehyde with alkynes. Reactions were performed reacting 2-azidobenzaldehyde with a range of terminal alkynes using 10 mol % of copper iodide as a catalyst, 20 mol % of thiourea as a ligand, triethylamine as base, DMSO as solvent at 100 °C under nitrogen atmosphere. The corresponding 2-(1H-1,2,3-triazoyl)-benzaldehydes (2-TBH) were obtained in moderated to excellent yields and according our experiments, the use of thiourea decreases the formation of side products. The obtained compounds were screened for their binding affinity with multiple therapeutic targets of AD by molecular docking: β-secretase (BACE), glycogen synthase kinase (GSK-3β) and acetylcholinesterase (AChE). The three compounds with highest affinity, 5 a (2-(4-phenyl-1H-1,2,3-triazol-1-yl)benzaldehyde), 5 b (2-(4-(p-tolyl)-1H-1,2,3-triazol-1-yl)benzaldehyde), and 5 d (2-(4-(4-(tert-butyl)phenyl)-1H-1,2,3-triazol-1-yl)benzaldehyde) were selected and evaluated on its antioxidant effect, in view of select the most promising one to perform the in vivo validation. Due the antioxidant potential ally to the affinity with BACE, GSK-3β and AChE, compound 5 b was evaluated in a mouse model of AD induced by intracerebroventricular injection of streptozotocin (STZ). Our results indicate that 5 b (1 mg/kg) treatment during 20 days is able to reverse the cognitive and memory impairment induced by STZ trough the modulation of AChE activity, amyloid cascade and GSK-3β expression.
Collapse
Affiliation(s)
- Gabriel P Costa
- LASOL-CCQFA, Universidade Federal de Pelotas - UFPel, P.O. Box 354 - 96010-900, Pelotas RS, Brazil
| | - Rodolfo S M Baldinotti
- Grupo de Pesquisa em Neurobiotecnologia - GPN, CDTec, Universidade Federal de Pelotas, Pelotas RS, Brazil
| | - Mariana G Fronza
- Grupo de Pesquisa em Neurobiotecnologia - GPN, CDTec, Universidade Federal de Pelotas, Pelotas RS, Brazil
| | | | - Ítalo F C Dias
- LASOL-CCQFA, Universidade Federal de Pelotas - UFPel, P.O. Box 354 - 96010-900, Pelotas RS, Brazil
| | - Mariana Souza Sonego
- Grupo de Pesquisa em Oncologia, Universidade Federal de Pelotas, Pelotas RS, Brazil
| | | | - Tiago Collares
- Grupo de Pesquisa em Oncologia, Universidade Federal de Pelotas, Pelotas RS, Brazil
| | - Gelson Perin
- LASOL-CCQFA, Universidade Federal de Pelotas - UFPel, P.O. Box 354 - 96010-900, Pelotas RS, Brazil
| | - Raquel G Jacob
- LASOL-CCQFA, Universidade Federal de Pelotas - UFPel, P.O. Box 354 - 96010-900, Pelotas RS, Brazil
| | - Lucielli Savegnago
- Grupo de Pesquisa em Neurobiotecnologia - GPN, CDTec, Universidade Federal de Pelotas, Pelotas RS, Brazil
| | - Diego Alves
- LASOL-CCQFA, Universidade Federal de Pelotas - UFPel, P.O. Box 354 - 96010-900, Pelotas RS, Brazil
| |
Collapse
|
13
|
Liang S, Wang Z, Yuan J, Zhang J, Dai X, Qin F, Zhang J, Sun Y. Rapid Identification of Tanshinone IIA Metabolites in an Amyloid-β 1-42 Induced Alzherimer's Disease Rat Model using UHPLC-Q-Exactive Qrbitrap Mass Spectrometry. Molecules 2019; 24:molecules24142584. [PMID: 31315255 PMCID: PMC6680413 DOI: 10.3390/molecules24142584] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 07/08/2019] [Accepted: 07/09/2019] [Indexed: 01/21/2023] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disorder that damages health and welfare of the elderly, and there has been no effective therapy for AD until now. It has been proved that tanshinone IIA (tan IIA) could alleviate pathological symptoms of AD via improving non-amyloidogenic cleavage of amyloid precursor protein, decreasing the accumulations of p-tau and amyloid-β1–42 (Aβ1–42), and so forth. However, the further biochemical mechanisms of tan IIA are not clear. The experiment was undertaken to explore metabolites of tan IIA in AD rats induced by microinjecting Aβ1-42 in the CA1 region of hippocampus. AD rats were orally administrated with tan IIA at 100 mg/kg weight, and plasma, urine, faeces, kidney, liver and brain were then collected for metabolites analysis by UHPLC-Q-Exactive Qrbitrap mass spectrometry. Consequently, a total of 37 metabolites were positively or putatively identified on the basis of mass fragmentation behavior, accurate mass measurements and retention times. As a result, methylation, hydroxylation, dehydration, decarbonylation, reduction reaction, glucuronidation, glycine linking and their composite reactions were characterized to illuminate metabolic pathways of tan IIA in vivo. Several metabolites presented differences in the distribution of tan IIA between the sham control and the AD model group. Overall, these results provided valuable references for research on metabolites of tan IIA in vivo and its probable active structure for exerting neuroprotection.
Collapse
Affiliation(s)
- Shuang Liang
- College of Biochemical Engineering, Beijing Union University, Beijing 100191, China
| | - Zijian Wang
- Beijing University of Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing 100191, China
| | - Jiaqi Yuan
- College of Biochemical Engineering, Beijing Union University, Beijing 100191, China
| | - Jing Zhang
- College of Biochemical Engineering, Beijing Union University, Beijing 100191, China
| | - Xueling Dai
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing 100191, China
| | - Fei Qin
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing 100191, China
| | - Jiayu Zhang
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China
| | - Yaxuan Sun
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing 100191, China.
| |
Collapse
|
14
|
Takano A, Chen L, Nag S, Brodney MA, Arakawa R, Chang C, Amini N, Doran SD, Dutra JK, McCarthy TJ, Nolan CE, O'Neill BT, Villalobos A, Zhang L, Halldin C. Quantitative Analysis of 18F-PF-06684511, a Novel PET Radioligand for Selective β-Secretase 1 Imaging, in Nonhuman Primate Brain. J Nucl Med 2018; 60:992-997. [PMID: 30530832 DOI: 10.2967/jnumed.118.217372] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 11/26/2018] [Indexed: 11/16/2022] Open
Abstract
β-secretase 1 (BACE1) is a key enzyme in the generation of β-amyloid, which is accumulated in the brain of Alzheimer disease patients. PF-06684511 was identified as a candidate PET ligand for imaging BACE1 in the brain and showed high specific binding in an initial assessment in a nonhuman primate (NHP) PET study using 18F-PF-06684511. In this effort, we aimed to quantitatively evaluate the regional brain distribution of 18F-PF-06684511 in NHPs under baseline and blocking conditions and to assess the target occupancy of BACE1 inhibitors. In addition, NHP whole-body PET measurements were performed to estimate the effective radiation dose. Methods: Initial brain PET measurements were performed at baseline and after oral administration of 5 mg/kg of LY2886721, a BACE1 inhibitor, in 2 cynomolgus monkeys. Kinetic analysis was performed with the radiometabolite-corrected plasma input function. In addition, a wide dose range of another BACE1 inhibitor, PF-06663195, was examined to investigate the relationship between the brain target occupancy and plasma concentration of the drug. Finally, the effective radiation dose of 18F-PF-06684511 was estimated on the basis of the whole-body PET measurements in NHPs. Results: Radiolabeling was accomplished successfully with an incorporation radiochemical yield of 4%-12% (decay-corrected) from 18F ion. The radiochemical purity was greater than 99%. The whole-brain uptake of 18F-PF-06684511 peaked (∼220% SUV) at approximately 20 min and decreased thereafter (∼100% SUV at 180 min). A 2-tissue-compartment model described the time-activity curves well. Pretreatment with LY2886721 reduced the total distribution volume of 18F-PF-06684511 by 48%-80% depending on the brain region, confirming its in vivo specificity. BACE1 occupancy of PF-06663195, estimated using the Lassen occupancy plot, showed a dose-dependent increase. The effective dose of 18F-PF-06684511 was 0.043 mSv/MBq for humans. Conclusion: 18F-PF-06684511 is the first successful PET radioligand for BACE1 brain imaging that demonstrates favorable in vivo binding and brain kinetics in NHPs.
Collapse
Affiliation(s)
- Akihiro Takano
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Laigao Chen
- Worldwide Research and Development, Pfizer Inc., Cambridge, Massachusetts; and
| | - Sangram Nag
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Michael A Brodney
- Worldwide Research and Development, Pfizer Inc., Cambridge, Massachusetts; and
| | - Ryosuke Arakawa
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Cheng Chang
- Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - Nahid Amini
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Shawn D Doran
- Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - Jason K Dutra
- Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - Timothy J McCarthy
- Worldwide Research and Development, Pfizer Inc., Cambridge, Massachusetts; and
| | - Charles E Nolan
- Worldwide Research and Development, Pfizer Inc., Cambridge, Massachusetts; and
| | - Brian T O'Neill
- Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | | | - Lei Zhang
- Worldwide Research and Development, Pfizer Inc., Cambridge, Massachusetts; and
| | - Christer Halldin
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| |
Collapse
|
15
|
Cao Q, Shin WS, Chan H, Vuong CK, Dubois B, Li B, Murray KA, Sawaya MR, Feigon J, Black DL, Eisenberg DS, Jiang L. Inhibiting amyloid-β cytotoxicity through its interaction with the cell surface receptor LilrB2 by structure-based design. Nat Chem 2018; 10:1213-1221. [PMID: 30297750 PMCID: PMC6250578 DOI: 10.1038/s41557-018-0147-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 08/28/2018] [Indexed: 12/31/2022]
Abstract
Inhibiting the interaction between amyloid-β (Aβ) and a neuronal cell surface receptor, LilrB2, has been suggested as a potential route for treating Alzheimer's disease. Supporting this approach, Alzheimer's-like symptoms are reduced in mouse models following genetic depletion of the LilrB2 homologue. In its pathogenic, oligomeric state, Aβ binds to LilrB2, triggering a pathway to synaptic loss. Here we identify the LilrB2 binding moieties of Aβ (16KLVFFA21) and identify its binding site on LilrB2 from a crystal structure of LilrB2 immunoglobulin domains D1D2 complexed to small molecules that mimic phenylalanine residues. In this structure, we observed two pockets that can accommodate the phenylalanine side chains of KLVFFA. These pockets were confirmed to be 16KLVFFA21 binding sites by mutagenesis. Rosetta docking revealed a plausible geometry for the Aβ-LilrB2 complex and assisted with the structure-guided selection of small molecule inhibitors. These molecules inhibit Aβ-LilrB2 interactions in vitro and on the cell surface and reduce Aβ cytotoxicity, which suggests these inhibitors are potential therapeutic leads against Alzheimer's disease.
Collapse
Affiliation(s)
- Qin Cao
- Departments of Chemistry and Biochemistry and Biological Chemistry, UCLA-DOE Institute and Howard Hughes Medical Institute, UCLA, Los Angeles, CA, USA
| | - Woo Shik Shin
- Department of Neurology, Molecular Biology Institute and Brain Research Institute, UCLA, Los Angeles, CA, USA
| | - Henry Chan
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA, USA
| | - Celine K Vuong
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
- Systems Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Bethany Dubois
- Department of Neurology, Molecular Biology Institute and Brain Research Institute, UCLA, Los Angeles, CA, USA
- Division of Applied Mathematics, Brown University, Providence, RI, USA
| | - Binsen Li
- Department of Neurology, Molecular Biology Institute and Brain Research Institute, UCLA, Los Angeles, CA, USA
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA, USA
| | - Kevin A Murray
- Departments of Chemistry and Biochemistry and Biological Chemistry, UCLA-DOE Institute and Howard Hughes Medical Institute, UCLA, Los Angeles, CA, USA
| | - Michael R Sawaya
- Departments of Chemistry and Biochemistry and Biological Chemistry, UCLA-DOE Institute and Howard Hughes Medical Institute, UCLA, Los Angeles, CA, USA
| | - Juli Feigon
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA, USA
| | - Douglas L Black
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - David S Eisenberg
- Departments of Chemistry and Biochemistry and Biological Chemistry, UCLA-DOE Institute and Howard Hughes Medical Institute, UCLA, Los Angeles, CA, USA.
| | - Lin Jiang
- Department of Neurology, Molecular Biology Institute and Brain Research Institute, UCLA, Los Angeles, CA, USA.
| |
Collapse
|
16
|
Pradhan N, Debnath K, Mandal S, Jana NR, Jana NR. Antiamyloidogenic Chemical/Biochemical-Based Designed Nanoparticle as Artificial Chaperone for Efficient Inhibition of Protein Aggregation. Biomacromolecules 2018; 19:1721-1731. [DOI: 10.1021/acs.biomac.8b00671] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Nibedita Pradhan
- Centre for Advanced Materials, Indian Association for the Cultivation of Science, Kolkata 700032, India
| | - Koushik Debnath
- Centre for Advanced Materials, Indian Association for the Cultivation of Science, Kolkata 700032, India
| | - Suman Mandal
- Centre for Advanced Materials, Indian Association for the Cultivation of Science, Kolkata 700032, India
| | - Nihar R. Jana
- Cellular and Molecular Neuroscience Laboratory, National Brain Research Centre, Manesar, Gurgaon 122051, India
| | - Nikhil R. Jana
- Centre for Advanced Materials, Indian Association for the Cultivation of Science, Kolkata 700032, India
| |
Collapse
|
17
|
Wang D, Schultz T, Novak GP, Baker S, Bennett DA, Narayan VA. Longitudinal Modeling of Functional Decline Associated with Pathologic Alzheimer's Disease in Older Persons without Cognitive Impairment. J Alzheimers Dis 2018; 62:855-865. [PMID: 29480187 PMCID: PMC6195203 DOI: 10.3233/jad-170903] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Therapeutic research on Alzheimer's disease (AD) has moved to intercepting the disease at the preclinical phase. Most drugs in late development have focused on the amyloid hypothesis. OBJECTIVE To understand the magnitude of amyloid-related functional decline and to identify the functional domains sensitive to decline in a preclinical AD population. METHODS Data were from the Religious Orders Study and the Rush Memory and Aging Project. Cognitive decline was measured by a modified version of the Alzheimer's Disease Cooperative Study Preclinical Alzheimer Cognitive Composite. The trajectories of functional decline, as measured by the instrumental and basic activities of daily living, were longitudinally modeled in 484 participants without cognitive impairment at baseline and having both a final clinical and a postmortem neuropathology assessment of AD. RESULTS Individuals with different final clinical diagnoses had different trajectories of cognitive and functional decline. Individuals with AD dementia, minor cognitive impairment, and no cognitive impairment had the most, intermediate, and least declines. While individuals with pathologic AD had significantly more cognitive decline over time than those without, the magnitude of difference in functional decline between these two groups was small. Functional domains such as handling finance and handling medications were more sensitive to decline. CONCLUSION Demonstrating the functional benefit of an amyloid-targeting drug represents a significant challenge as elderly people experience functional decline due to a wide range of reasons with limited manifestation attributable to AD neuropathology. More sensitive functional scales focusing on the functional domains sensitive to decline in preclinical AD are needed.
Collapse
Affiliation(s)
- Dai Wang
- R&D Information Technology, Janssen Research & Development, LLC, Titusville, NJ, USA
- Neuroscience Therapeutic Area, Janssen Research & Development, LLC, Titusville, NJ, USA
| | - Tim Schultz
- R&D Information Technology, Janssen Research & Development, LLC, Titusville, NJ, USA
- Neuroscience Therapeutic Area, Janssen Research & Development, LLC, Titusville, NJ, USA
| | - Gerald P. Novak
- Neuroscience Therapeutic Area, Janssen Research & Development, LLC, Titusville, NJ, USA
| | - Susan Baker
- Neuroscience Therapeutic Area, Janssen Research & Development, LLC, Titusville, NJ, USA
| | - David A. Bennett
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Vaibhav A. Narayan
- R&D Information Technology, Janssen Research & Development, LLC, Titusville, NJ, USA
- Neuroscience Therapeutic Area, Janssen Research & Development, LLC, Titusville, NJ, USA
| |
Collapse
|
18
|
Subramanian G, Poda G. In silico ligand-based modeling of hBACE-1 inhibitors. Chem Biol Drug Des 2017; 91:817-827. [PMID: 29139199 DOI: 10.1111/cbdd.13147] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 10/24/2017] [Accepted: 11/01/2017] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease is a chronic neurodegenerative disease affecting more than 30 million people worldwide. Development of small molecule inhibitors of human β-secretase 1 (hBACE-1) is being the focus of pharmaceutical industry for the past 15-20 years. Here, we successfully applied multiple ligand-based in silico modeling techniques to understand the inhibitory activities of a diverse set of small molecule hBACE-1 inhibitors reported in the scientific literature. Strikingly, the use of only a small subset of 230 (13%) molecules allowed us to develop quality models that performed reasonably well on the validation set of 1,476 (87%) inhibitors. Varying the descriptor sets and the complexity of the modeling techniques resulted in only minor improvements to the model's performance. The current results demonstrate that predictive models can be built by choosing appropriate modeling techniques in spite of using small datasets consisting of diverse chemical classes, a scenario typical in triaging of high-throughput screening results to identify false negatives. We hope that these encouraging results will help the community to develop more predictive models that would support research efforts for the debilitating Alzheimer's disease. Additionally, the integrated diversity of the techniques employed will stimulate scientists in the field to use in silico statistical modeling techniques like these to derive better models to help advance the drug discovery projects faster.
Collapse
Affiliation(s)
| | - Gennady Poda
- Drug Discovery Program, Ontario Institute for Cancer Research, Toronto, ON, Canada.,Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
19
|
Gueto-Tettay C, Pelaez-Bedoya L, Drosos-Ramirez JC. Population density analysis for determining the protonation state of the catalytic dyad in BACE1-tertiary carbinamine-based inhibitor complex. J Biomol Struct Dyn 2017; 36:3557-3574. [DOI: 10.1080/07391102.2017.1393461] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Carlos Gueto-Tettay
- Grupo de Química Bioorgánica, Universidad de Cartagena, Cartagena de Indias, Colombia
| | - Luis Pelaez-Bedoya
- Grupo de Química Bioorgánica, Universidad de Cartagena, Cartagena de Indias, Colombia
| | | |
Collapse
|
20
|
Identification and characterization of new isoforms of human fas apoptotic inhibitory molecule (FAIM). PLoS One 2017; 12:e0185327. [PMID: 28981531 PMCID: PMC5628826 DOI: 10.1371/journal.pone.0185327] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 09/11/2017] [Indexed: 01/07/2023] Open
Abstract
Fas Apoptosis Inhibitory Molecule (FAIM) is an evolutionarily highly conserved death receptor antagonist, widely expressed and known to participate in physiological and pathological processes. Two FAIM transcript variants have been characterized to date, namely FAIM short (FAIM-S) and FAIM long (FAIM-L). FAIM-S is ubiquitously expressed and serves as an anti-apoptotic protein in the immune system. Furthermore, in neurons, this isoform promotes NGF-induced neurite outgrowth through NF-кB and ERK signaling. In contrast FAIM-L is found only in neurons, where it exerts anti-apoptotic activity against several stimuli. In addition to these two variants, in silico studies point to the existence of two additional isoforms, neither of which have been characterized to date. In this regard, here we confirm the presence of these two additional FAIM isoforms in human fetal brain, fetal and adult testes, and placenta tissues. We named them FAIM-S_2a and FAIM-L_2a since they have the same sequence as FAIM-S and FAIM-L, but include exon 2a. PCR and western blot revealed that FAIM-S_2a shows ubiquitous expression in all the tissues and cellular models tested, while FAIM-L_2a is expressed exclusively in tissues of the nervous system. In addition, we found that, when overexpressed in non-neuronal cells, the splicing factor nSR100 induces the expression of the neuronal isoforms, thus identifying it as responsible for the generation of FAIM-L and FAIM-L_2a. Functionally, FAIM-S_2a and FAIM-L_2a increased neurite outgrowth in response to NGF stimulation in a neuronal model. This observation thus, supports the notion that these two isoforms are involved in neuronal differentiation. Furthermore, subcellular fractionation experiments revealed that, in contrast to FAIM-S and FAIM-L, FAIM-S_2a and FAIM-L_2a are able to localize to the nucleus, where they may have additional functions. In summary, here we report on two novel FAIM isoforms that may have relevant roles in the physiology and pathology of the nervous system.
Collapse
|
21
|
Potent and Selective BACE-1 Peptide Inhibitors Lower Brain Aβ Levels Mediated by Brain Shuttle Transport. EBioMedicine 2017; 24:76-92. [PMID: 28923680 PMCID: PMC5652008 DOI: 10.1016/j.ebiom.2017.09.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 08/01/2017] [Accepted: 09/05/2017] [Indexed: 12/31/2022] Open
Abstract
Therapeutic approaches to fight Alzheimer's disease include anti-Amyloidβ (Aβ) antibodies and secretase inhibitors. However, the blood-brain barrier (BBB) limits the brain exposure of biologics and the chemical space for small molecules to be BBB permeable. The Brain Shuttle (BS) technology is capable of shuttling large molecules into the brain. This allows for new types of therapeutic modalities engineered for optimal efficacy on the molecular target in the brain independent of brain penetrating properties. To this end, we designed BACE1 peptide inhibitors with varying lipid modifications with single-digit picomolar cellular potency. Secondly, we generated active-exosite peptides with structurally confirmed dual binding mode and improved potency. When fused to the BS via sortase coupling, these BACE1 inhibitors significantly reduced brain Aβ levels in mice after intravenous administration. In plasma, both BS and non-BS BACE1 inhibitor peptides induced a significant time- and dose-dependent decrease of Aβ. Our results demonstrate that the BS is essential for BACE1 peptide inhibitors to be efficacious in the brain and active-exosite design of BACE1 peptide inhibitors together with lipid modification may be of therapeutic relevance.
Collapse
|
22
|
Blass B. C 5-C 6 Carbocyclic-Fused Iminothiadiazine Dioxides as BACE Inhibitors, Their Compositions, and Their Use. ACS Med Chem Lett 2017; 8:786-788. [PMID: 28835787 PMCID: PMC5554906 DOI: 10.1021/acsmedchemlett.7b00268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Indexed: 11/30/2022] Open
Affiliation(s)
- Benjamin Blass
- Temple University School of Pharmacy, 3307 North Broad Street, Philadelphia, Pennsylvania 19140, United States
| |
Collapse
|
23
|
Gueto-Tettay C, Martinez-Consuegra A, Zuchniarz J, Gueto-Tettay LR, Drosos-Ramírez JC. A PM7 dynamic residue-ligand interactions energy landscape of the BACE1 inhibitory pathway by hydroxyethylamine compounds. Part I: The flap closure process. J Mol Graph Model 2017; 76:274-288. [PMID: 28746905 DOI: 10.1016/j.jmgm.2017.07.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 07/10/2017] [Accepted: 07/11/2017] [Indexed: 01/08/2023]
Abstract
BACE1 is an enzyme of scientific interest because it participates in the progression of Alzheimer's disease. Hydroxyethylamines (HEAs) are a family of compounds which exhibit inhibitory activity toward BACE1 at a nanomolar level, favorable pharmacokinetic properties and oral bioavailability. The first step in the inhibition of BACE1 by HEAs consists of their entrance into the protease active site and the resultant conformational change in the protein, from Apo to closed form. These two conformations differ in the position of an antiparallel loop (called the flap) which covers the entrance to the catalytic site. For BACE1, closure of this flap is vital to its catalytic activity and to inhibition of the enzyme due to the new interactions thereby formed with the ligand. In the present study a dynamic energy landscape of residue-ligand interaction energies (ReLIE) measured for 112 amino acids in the BACE1 active site and its immediate vicinity during the closure of the flap induced by 8 HEAs of different inhibitory power is presented. A total of 6.272 million ReLIE calculations, based on the PM7 semiempirical method, provided a deep and quantitative view of the first step in the inhibition of the aspartyl protease. The information suggests that residues Asp93, Asp289, Thr292, Thr293, Asn294 and Arg296 are anchor points for the ligand, accounting for approximately 45% of the total protein-ligand interaction. Additionally, flap closure improved the BACE1-HEA interaction by around 25%. Furthermore, the inhibitory activity of HEAs could be related to the capacity of these ligands to form said anchor point interactions and maintain them over time: the lack of some of these anchor interactions delayed flap closure or impeded it completely, or even caused the flap to reopen. The methodology employed here could be used as a tool to evaluate future structural modifications which lead to improvements in the favorability and stability of BACE1-HEA ReLIEs, aiding in the design of better inhibitors.
Collapse
Affiliation(s)
- Carlos Gueto-Tettay
- Grupo de Química Bioorgánica, Facultad de Ciencias Exactas y Naturales, Universidad de Cartagena, Cartagena, Colombia.
| | - Alejandro Martinez-Consuegra
- Grupo de Química Bioorgánica, Facultad de Ciencias Exactas y Naturales, Universidad de Cartagena, Cartagena, Colombia
| | - Joshua Zuchniarz
- Grupo de Química Bioorgánica, Facultad de Ciencias Exactas y Naturales, Universidad de Cartagena, Cartagena, Colombia
| | - Luis Roberto Gueto-Tettay
- Grupo de Química Bioorgánica, Facultad de Ciencias Exactas y Naturales, Universidad de Cartagena, Cartagena, Colombia
| | - Juan Carlos Drosos-Ramírez
- Grupo de Química Bioorgánica, Facultad de Ciencias Exactas y Naturales, Universidad de Cartagena, Cartagena, Colombia.
| |
Collapse
|
24
|
Alzheimer's disease drug development pipeline: 2017. ALZHEIMERS & DEMENTIA-TRANSLATIONAL RESEARCH & CLINICAL INTERVENTIONS 2017; 3:367-384. [PMID: 29067343 PMCID: PMC5651419 DOI: 10.1016/j.trci.2017.05.002] [Citation(s) in RCA: 276] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION There is an urgent need to develop new treatments for Alzheimer's disease (AD) and to understand the drug development process for new AD therapies. METHODS We assessed the agents in the AD pipeline as documented in clinicaltrials.gov for phase I, phase II, and phase III, accessed 1/5/2017. RESULTS There are 105 agents in the AD treatment development pipeline, of which 25 agents are in 29 trials in phase I, 52 agents are in 68 trials in phase II, and 28 agents are in 42 trials in phase III. Seventy percent of drugs in the AD pipeline are disease-modifying therapies (DMTs). Fourteen percent are symptomatic cognitive enhancers, and 13% are symptomatic agents addressing neuropsychiatric and behavioral changes (2% have undisclosed mechanisms). Most trials are sponsored by the biopharmaceutical industry. Trials include patients with preclinical AD (cognitively normal with biomarker evidence of AD), prodromal AD (mild cognitive symptoms and biomarker evidence of AD), and AD dementia. Biomarkers are included in many drug development programs particularly those for DMTs. Thirteen of 46 phase II DMT trials have amyloid imaging as an entry criterion, and 10 of 28 phase III trials incorporate amyloid imaging for diagnosis and entry. A large number of participants are needed for AD clinical trials; in total, 54,073 participants are required for trials spanning preclinical AD to AD dementia. When compared with the 2016 pipeline, there are eight new agents in phase I, 16 in phase II, and five in phase III. DISCUSSION The AD drug development pipeline has 105 agents divided among phase I, phase II, and phase III. The trials include a wide range of clinical trial populations, many mechanisms of action, and require a substantial number of clinical trial participants. Biomarkers are increasingly used in patient identification and as outcome measures, particularly in trials of DMTs.
Collapse
|
25
|
Diling C, Tianqiao Y, Jian Y, Chaoqun Z, Ou S, Yizhen X. Docking Studies and Biological Evaluation of a Potential β-Secretase Inhibitor of 3-Hydroxyhericenone F from Hericium erinaceus. Front Pharmacol 2017; 8:219. [PMID: 28553224 PMCID: PMC5427148 DOI: 10.3389/fphar.2017.00219] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 04/07/2017] [Indexed: 12/25/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common neurodegenerative disorder, affecting approximately more than 5% of the population worldwide over the age 65, annually. The incidence of AD is expected to be higher in the next 10 years. AD patients experience poor prognosis and as a consequence new drugs and therapeutic strategies are required in order to improve the clinical responses and outcomes of AD. The purpose of the present study was to screen a certain number of potential compounds from herbal sources and investigate their corresponding mode of action. In the present study, the learning and memory effects of ethanol:water (8:2) extracts from Hericium erinaceus were evaluated on a dementia rat model. The model was established by intraperitoneal injection of 100 mg/kg/d D-galactose in rats. The results indicated that the extracts can significantly ameliorate the learning and memory abilities. Specific active ingredients were screened in vivo assays and the results were combined with molecular docking studies. Potential receptor–ligand interactions on the BACE1-inhibitor namely, 3-Hydroxyhericenone F (3HF) were investigated. The isolation of a limited amount of 3HF from the fruit body of H. erinaceus by chemical separation was conducted, and the mode of action of this compound was verified in NaN3-induced PC12 cells. The cell-based assays demonstrated that 3HF can significantly down-regulate the expression of BACE1 (p < 0.01), while additional AD intracellular markers namely, p-Tau and Aβ1-42 were further down-regulated (p < 0.05). The data further indicate that 3HF can ameliorate certain mitochondrial dysfunction conditions by the reversal of the decreasing level of mitochondrial respiratory chain complexes, the calcium ion levels ([Ca2+]), the inhibiton in the production of ROS, the increase in the mitochondrial membrane potential and ATP levels, and the regulation of the expression levels of the genes encoding for the p21, COX I, COX II, PARP1, and NF-κB proteins. The observations suggest the use of H. erinaceus in traditional medicine for the treatment of various neurological diseases and render 3HF as a promising naturally occurring chemical constituent for the treatment of AD via the inhibition of the β-secretase enzyme.
Collapse
Affiliation(s)
- Chen Diling
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of MicrobiologyGuangzhou, China
| | - Yong Tianqiao
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of MicrobiologyGuangzhou, China
| | - Yang Jian
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of MicrobiologyGuangzhou, China
| | - Zheng Chaoqun
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of MicrobiologyGuangzhou, China.,College of Chinese Material Medical, Guangzhou University of Chinese MedicineGuangzhou, China
| | - Shuai Ou
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of MicrobiologyGuangzhou, China
| | - Xie Yizhen
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of MicrobiologyGuangzhou, China
| |
Collapse
|
26
|
Parsons CG, Rammes G. Preclinical to phase II amyloid beta (Aβ) peptide modulators under investigation for Alzheimer’s disease. Expert Opin Investig Drugs 2017; 26:579-592. [DOI: 10.1080/13543784.2017.1313832] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- Chris G. Parsons
- Non-Clinical Science, Merz Pharmaceuticals GmbH, Frankfurt am Main, Germany
| | - Gerhard Rammes
- Klinikum rechts der Isar der Technischen Universitat Munchen – Department of Anesthesiology, Munchen, Germany
| |
Collapse
|
27
|
Hao C, Wang W, Wang S, Zhang L, Guo Y. An Overview of the Protective Effects of Chitosan and Acetylated Chitosan Oligosaccharides against Neuronal Disorders. Mar Drugs 2017; 15:md15040089. [PMID: 28333077 PMCID: PMC5408235 DOI: 10.3390/md15040089] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 03/07/2017] [Accepted: 03/15/2017] [Indexed: 12/12/2022] Open
Abstract
Chitin is the second most abundant biopolymer on Earth and is mainly comprised of a marine invertebrate, consisting of repeating β-1,4 linked N-acetylated glucosamine units, whereas its N-deacetylated product, chitosan, has broad medical applications. Interestingly, chitosan oligosaccharides have therapeutic effects on different types of neuronal disorders, including, but not limited to, Alzheimer’s disease, Parkinson’s disease, and nerve crush injury. A common link among neuronal disorders is observed at a sub-cellular level, such as atypical protein assemblies and induced neuronal death. Chronic activation of innate immune responses that lead to neuronal injury is also common in these diseases. Thus, the common mechanisms of neuronal disorders might explain the general therapeutic effects of chitosan oligosaccharides and their derivatives in these diseases. This review provides an update on the pathogenesis and therapy for neuronal disorders and will be mainly focused on the recent progress made towards the neuroprotective properties of chitosan and acetylated chitosan oligosaccharides. Their structural features and the underlying molecular mechanisms will also be discussed.
Collapse
Affiliation(s)
- Cui Hao
- Institute of Cerebrovascular Diseases, Affiliated Hospital of Qingdao University, Qingdao 266003, China.
| | - Wei Wang
- Key Laboratory of Marine Drugs, Ministry of Education, Ocean University of China, Qingdao 266003, China.
| | - Shuyao Wang
- Key Laboratory of Marine Drugs, Ministry of Education, Ocean University of China, Qingdao 266003, China.
| | - Lijuan Zhang
- Institute of Cerebrovascular Diseases, Affiliated Hospital of Qingdao University, Qingdao 266003, China.
| | - Yunliang Guo
- Institute of Cerebrovascular Diseases, Affiliated Hospital of Qingdao University, Qingdao 266003, China.
| |
Collapse
|
28
|
Allison B, Mani NS. Gram-Scale Synthesis of a β-Secretase 1 (BACE 1) Inhibitor. ACS OMEGA 2017; 2:397-408. [PMID: 30023606 PMCID: PMC6044763 DOI: 10.1021/acsomega.6b00362] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 01/25/2017] [Indexed: 06/08/2023]
Abstract
Development of a scalable synthesis of an oxazine class of β-secretase inhibitor is described. Trifluoromethylated acyloin synthesis by the reaction of a mandelic acid with trifluoroacetic anhydride in the presence of pyridine (Dakin-West reaction) was used as an efficient strategy to install the key trifluoromethyl substituent on the oxazine ring. Diastereoselective addition of methyl magnesium bromide to a cyclic sulfamidate imine and trimethylsilyl trifluoromethanesulfonate catalyzed intramolecular amidine formation to yield oxazine-3-amine are some of the significant, novel synthetic methods developed in this synthesis. These critical transformations allowed a concise 11-step route to the target compound with excellent overall yields.
Collapse
|
29
|
Ruthirakuhan M, Herrmann N, Suridjan I, Abraham EH, Farber I, Lanctôt KL. Beyond immunotherapy: new approaches for disease modifying treatments for early Alzheimer’s disease. Expert Opin Pharmacother 2016; 17:2417-2429. [DOI: 10.1080/14656566.2016.1258060] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
30
|
Dotsenko VV, Chigorina EA, Krivokolysko SG. 2,2,2-Trifluoroethyl-thiadiazines: a patent evaluation of WO2016023927. Expert Opin Ther Pat 2016; 26:1371-1376. [PMID: 27785946 DOI: 10.1080/13543776.2016.1254193] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
The Alzheimer's disease (AD) is acknowledged as the most common type of dementia in aging adults. It is characterized by the formation of intracellular neurofibrillary tangles and extracellular amyloid plaques. The latter insoluble deposits mainly consist of β-amyloid peptides (Aβ), which are the derivatives of the amyloid precursor protein (APP). The formation of neurotoxic Aβ-peptides involves the cleavage of APP with beta-secretase enzyme (beta-site APP cleaving enzyme 1, BACE1) so the potential of BACE1 inhibitors as therapeutic agents for AD is now drawing much attention. The patent application WO2016023927 reports the preparation of new 1,2,4-thiadiazine inhibitors of BACE1 activity and their use as therapeutically active substances. Some of the new compounds are claimed to be good inhibitors with the IC50 values in the 0.000292-0.134165 μM range. Several pharmaceutical preparations based on these compounds are proposed for possible treatment and prevention of AD. Expert opinion: In light of the novelty from the chemical point of view and improved biological activity, the reported 2,2,2-trifluoroethylthiadiazines could be considered as promising BACE1 inhibitors. However, the available data are insufficient to make a recommendation if these compounds can be considered as drug candidates. Further studies with a larger number of compounds are required. The compounds described in the patent have to be characterized more thoroughly from the chemical viewpoint (e.g., by means of IR, 1H and 13C NMR spectroscopy, X-ray crystallography), especially as regards stereochemical details.
Collapse
Affiliation(s)
- Victor V Dotsenko
- a Kuban State University , Krasnodar , Russia.,b ChemEx Lab, Vladimir Dal' Lugansk University , Lugansk , Russia
| | - Elena A Chigorina
- c Federal State Unitary Enterprise "State Scientific Research Institute of Chemical Reagents and High Purity Chemical Substances" (FSUE "IREA") , Moscow , Russia
| | - Sergey G Krivokolysko
- b ChemEx Lab, Vladimir Dal' Lugansk University , Lugansk , Russia.,d Lugansk State Medical University , Lugansk , Russia
| |
Collapse
|
31
|
Gueto-Tettay C, Zuchniarz J, Fortich-Seca Y, Gueto-Tettay LR, Drosos-Ramirez JC. A molecular dynamics study of the BACE1 conformational change from Apo to closed form induced by hydroxyethylamine derived compounds. J Mol Graph Model 2016; 70:181-195. [PMID: 27750187 DOI: 10.1016/j.jmgm.2016.10.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 10/05/2016] [Accepted: 10/06/2016] [Indexed: 12/11/2022]
Abstract
BACE1 is an aspartyl protease which is a therapeutic target for Alzheimer's disease (AD) because of its participation in the rate-limiting step in the production of Aβ-peptide, the accumulation of which produces senile plaques and, in turn, the neurodegenerative effects associated with AD. The active site of this protease is composed in part by two aspartic residues (Asp93 and Asp289). Additionally, the catalytic site has been found to be covered by an antiparallel hairpin loop called the flap. The dynamics of this flap are fundamental to the catalytic function of the enzyme. When BACE1 is inactive (Apo), the flap adopts an open conformation, allowing a substrate or inhibitor to access the active site. Subsequent interaction with the ligand induces flap closure and the stabilization of the macromolecular complex. Further, the protonation state of the aspartic dyad is affected by the chemical nature of the species entering the active site, so that appropriate selection of protonation states for the ligand and the catalytic residues will permit the elucidation of the inhibitory pathway for BACE1. In the present study, comparative analysis of different combinations of protonation states for the BACE1-hydroxyethylamine (HEA) system is reported. HEAs are potent inhibitors of BACE1 with favorable pharmacological and kinetic properties, as well as oral bioavailability. The results of Molecular Dynamics (MD) simulations and population density calculations using 8 different parameters demonstrate that the LnAsp289 configuration (HEA with a neutral amine and the Asp289 residue protonated) is the only one which permits the expected conformational change in BACE1, from apo to closed form, after flap closure. Additionally, differences in their capacities to establish and maintain interactions with residues such as Asp93, Gly95, Thr133, Asp289, Gly291, and Asn294 during this step allow differentiation among the inhibitory activities of the HEAs. The results and methodology here reported will serve to elucidate the inhibitory pathway of other families of compounds that act as BACE1 inhibitors, as well as the design of better leader compounds for the treatment of AD.
Collapse
Affiliation(s)
- Carlos Gueto-Tettay
- Grupo de Química Bioorgánica, Facultad de Ciencias Exactas y Naturales, Universidad de Cartagena, Colombia
| | - Joshua Zuchniarz
- Grupo de Química Bioorgánica, Facultad de Ciencias Exactas y Naturales, Universidad de Cartagena, Colombia
| | - Yeyson Fortich-Seca
- Grupo de Química Bioorgánica, Facultad de Ciencias Exactas y Naturales, Universidad de Cartagena, Colombia
| | - Luis Roberto Gueto-Tettay
- Grupo de Química Bioorgánica, Facultad de Ciencias Exactas y Naturales, Universidad de Cartagena, Colombia
| | - Juan Carlos Drosos-Ramirez
- Grupo de Química Bioorgánica, Facultad de Ciencias Exactas y Naturales, Universidad de Cartagena, Colombia.
| |
Collapse
|